1
|
Zeng H, Feng H, Zhang C, Kang Z, Wu J, Zhao X, Huang A, Xu Y, Huang Y, Xu H, Gong M. Novel intravenous formulation for radiosensitization in osteosarcoma treatment. Mater Today Bio 2025; 32:101682. [PMID: 40206141 PMCID: PMC11979400 DOI: 10.1016/j.mtbio.2025.101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents. While radiotherapy is an adjuvant treatment option for OS, particularly in cases of unresectable recurrent metastases, its efficacy remains limited. Enhancing radiosensitivity in OS cells is therefore crucial for improving treatment outcomes. Hafnium oxide, a known radiosensitizer, has demonstrated potential but its current formulation restricts its use to intratumoral administration, posing challenges for treating intraosseous tumors. The development of an intravenous formulation is thus highly desirable. Furthermore, radiotherapy resistance, driven by tumor hypoxia and an immunosuppressive microenvironment, further compromises its effectiveness. In this study, we synthesized hafnium-doped Prussian blue nanoparticles (HP) coated with a tannic acid-manganese metallophenol network (HPTM) to improve biocompatibility and enable intravenous administration. Following intravenous injection in a murine model of OS tibialis in situ tumors with lung metastases, HPTM effectively localized to the primary tumor. Within the acidic tumor microenvironment, manganese was released, activating the STING pathway and triggering anti-tumor immune responses. Moreover, near-infrared light irradiation of the Prussian blue component induced a photothermal effect, promoting apoptosis. Concurrently, under low-dose X-ray irradiation, HPTM augmented radiation energy deposition, generating reactive oxygen species and inducing DNA damage in tumor cells. This synergistic therapeutic approach significantly increased apoptosis in radiotherapy-resistant OS cells, reduced lung metastases, and suppressed primary tumor growth. These findings suggest a promising avenue for clinical translation, integrating radiosensitization, photothermal therapy, and STING pathway activation to overcome current limitations in OS radiotherapy.
Collapse
Affiliation(s)
- Haitao Zeng
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
- Bone&Soft Tissue Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430079, China
| | - Huixiong Feng
- Department of Musculoskeletal Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chong Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Zhe Kang
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
| | - Jianping Wu
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
| | - Xingqi Zhao
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
| | - Anfei Huang
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
| | - Yanyang Xu
- Department of Joint Surgery, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Yufeng Huang
- Department of Cervical Spondylosis and Spine Orthopedics, The First Af Liated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, China
| | - Hongwen Xu
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
| | - Ming Gong
- Department of Pediatric Orthopaedics, GuangZhou Women and Children's Medical Center, GuangZhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, GuangZhou, 510623, China
| |
Collapse
|
2
|
Wang L, Gao Z, Tian M, Liu L, Xie J, Chen M, Huang Z, Dong B, Li W, Shi L, Tong Y, Xu H, Shen B, Cen D, Yu H, Yu X. A Nanosystem Alleviates Severe Acute Pancreatitis via Reactive Oxygen Species Scavenging and Enhancing Mitochondrial Autophagy. NANO LETTERS 2025; 25:8644-8654. [PMID: 40369909 PMCID: PMC12123669 DOI: 10.1021/acs.nanolett.5c01495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Severe acute pancreatitis (SAP) is a life-threatening condition characterized by excessive reactive oxygen species (ROS) production and impaired mitochondrial function, resulting from disrupted autophagic flux. Current clinical treatment for SAP fails to address the condition comprehensively, with the treatment targeting only a single pathogenesis. Herein, we report an innovative acid-responsive biomimetic nanozyme. This system features a hollow Prussian blue (PB) core, serving as an ROS scavenger encapsulated within a porous ZIF-8 shell, enabling the efficient delivery of celastrol that activates autophagic flux. Encased in a macrophage membrane, this system selectively targets inflamed pancreatic tissues and is readily internalized by pancreatic acinar cells. This dual-scavenging mechanism effectively attenuates inflammatory cytokine levels and restores mitochondrial homeostasis in three distinct SAP mouse models. Overall, this study presents a promising synergistic strategy for the dual scavenging of damaged mitochondria and ROS, offering a novel therapeutic approach to the treatment of SAP.
Collapse
Affiliation(s)
- Liying Wang
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Zerui Gao
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Mengxiang Tian
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Li Liu
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Jinyan Xie
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Muxiong Chen
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Zihao Huang
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Bingzhi Dong
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Weiqi Li
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Liang Shi
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
| | - Yifan Tong
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Hongxia Xu
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
| | - Bo Shen
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Dong Cen
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
| | - Hong Yu
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| | - Xin Yu
- Department
of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic
of China
- Zhejiang
Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection,
Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
- Department
of Anesthesia, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang310016, People’s Republic of China
| |
Collapse
|
3
|
Khan M, Ullah R, Shah SM, Farooq U, Li J. Manganese-Based Nanotherapeutics for Targeted Treatment of Breast Cancer. ACS APPLIED BIO MATERIALS 2025; 8:3571-3600. [PMID: 40293195 DOI: 10.1021/acsabm.5c00040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Breast cancer (BC) is one of the most common cancers among women and is associated with high mortality. Traditional modalities, including surgery, radiotherapy, and chemotherapy, have achieved certain advancements but continue to combat challenges including harm to healthy tissues, resistance to treatment, and adverse drug reactions. The rapid advancements in nanotechnology recently facilitated the exploration of innovative strategies for breast cancer therapy. Manganese-based nanotherapeutics have attracted great attention because of their unique characteristics such as tunable structures/morphologies, versatility, magnetic/optical properties, strong catalytic activities, excellent biodegradability, and biocompatibility. In this review, we highlighted different types of Mn-based nanotherapeutics to modulate TME, including metal-immunotherapy, alleviating tumor hypoxia, and increasing reactive oxygen species production, and we emphasized its role in magnetic resonance imaging (MRI)-guided therapy, photoacoustic imaging, and theranostic-based therapy along with a therapeutic carrier, all of which were discussed in the context of breast cancer. Hopefully, the present review will provide insights into the current landscape and future directions of multifunctional applications of Mn-based nanotherapeutics in the field of breast cancer treatment.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, P.R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants, College of Bioengineering, Chongqing University, Chongqing 400030, China
- Jinfeng Laboratory, No. 313 Jinyue Road, High-tech Zone, Chongqing 401329, China
| | - Syed Mubassir Shah
- Department of Biotechnology, Abdul Wali Khan University, KPK, Mardan 23200, Pakistan
| | - Umar Farooq
- Jinfeng Laboratory, No. 313 Jinyue Road, High-tech Zone, Chongqing 401329, China
| | - Jun Li
- Jinfeng Laboratory, No. 313 Jinyue Road, High-tech Zone, Chongqing 401329, China
| |
Collapse
|
4
|
Li X, Xu S, Su Z, Shao Z, Huang X. Unleashing the Potential of Metal Ions in cGAS-STING Activation: Advancing Nanomaterial-Based Tumor Immunotherapy. ACS OMEGA 2025; 10:11723-11742. [PMID: 40191377 PMCID: PMC11966298 DOI: 10.1021/acsomega.4c10865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 04/09/2025]
Abstract
Immunotherapy is a critical modality in cancer treatment with diverse activation pathways. In recent years, the stimulator of interferon genes (STING) signaling pathway has exhibited significant potential in tumor immunotherapy. This pathway exerts notable antitumor effects by activating innate and adaptive immunity and regulating the tumor immune microenvironment. Various metal ions have been identified as effective activators of the STING pathway and, through the design and synthesis of nanodelivery platforms, have been applied in immunotherapy as well as in combination therapies, such as chemotherapy, chemodynamic therapy, photodynamic therapy, and cancer vaccines. Metal nanomaterials showcase unique advantages in immunotherapy; however, there are still aspects that require optimization. This review systematically examines existing metal-based nanomaterials, elaborates on the mechanisms by which different metal ions activate the STING pathway, and discusses their application models in tumor combination therapies. We also provide a comparative analysis of the advantages of metal nanomaterials over other treatment methods. Our exploration highlights the broad application prospects of metal nanomaterials in cancer treatment, offering new insights and directions for the advancement of tumor immunotherapy.
Collapse
Affiliation(s)
- Xingyin Li
- Department
of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shaojie Xu
- Department
of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ziliang Su
- Department
of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zengwu Shao
- Department
of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Huang
- Department
of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
5
|
Hu Y, Qi E, Yun C, Li X, Liu F, Cheng Z, Guan N, Wang Q, Zhao H, Xiao W, Peng L, Yang J, Yu X. Photothermal therapy combined with a STING agonist induces pyroptosis, and gasdermin D could be a new biomarker for guiding the treatment of pancreatic cancer. J Transl Med 2025; 23:271. [PMID: 40038726 PMCID: PMC11877846 DOI: 10.1186/s12967-025-06247-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/11/2025] [Indexed: 03/06/2025] Open
Abstract
PURPOSE Although photothermal therapy (PTT) can induce antitumour immunity, the mechanisms underlying its effects in pancreatic cancer (PC) require further exploration. In this study, the mechanism of action of PTT and its connection to pyroptosis as well as the therapeutic potential of PTT alone and in combination with STING agonists, were investigated. In addition, a biomarker of PC was found to stratify patients who are suitable for PTT. EXPERIMENTAL DESIGN We explored whether PTT can induce pyroptosis in vitro and evaluated the therapeutic efficacy and antitumour immunity-inducing ability of PTT combined with STING agonist (c-di-GMP) as immune adjuvant in vivo in PC. We also evaluated gasdermin D (GSDMD) expression in tumour tissues and investigated drug sensitivity in patient-derived organoids (PDOs) with differential GSDMD expression. RESULTS Our study demonstrated that local PTT induces pyroptosis via the caspase-1/GSDMD pathway and elicits antitumour immunity. PTT combined with a STING agonist exhibits better therapeutic efficacy than PTT alone while limiting distant tumour metastasis, and enhances the immune response by promoting dendritic cell maturation, increasing the frequency of tumour infiltrating T cells, and converting macrophages from the M2 to the M1 phenotype. In addition, we found that GSDMD is highly expressed in tumour tissues and that overexpression of GSDMD in PC might suggest increased resistance to chemotherapy and the potential benefits of local therapy. We further confirmed that PDOs with higher GSDMD expression are less sensitive to a chemotherapeutic agent (5-Fluorouracil) than PDOs with lower GSDMD expression, making GSDMD a new biomarker for identifying patients who may benefit from PTT. CONCLUSIONS In this work, c-di-GMP was used as an immune adjuvant for PTT to treat PC for the first time, and the results provide clues for the development of novel combination immunotherapies that simultaneously suppress primary tumours and distant metastases. GSDMD has great potential as a new biomarker for the selection of individualized treatment modalities.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - ErPeng Qi
- Department of Interventional Ultrasound, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Chao Yun
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Specialty in Oncology, Jinzhou Medical University, Jinzhou, China
| | - Xi Li
- Department of Urology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation, Cambridge, UK
| | - Fangyi Liu
- Department of Interventional Ultrasound, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Zhigang Cheng
- Department of Interventional Ultrasound, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China
| | - Na Guan
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China
- Specialty in Oncology, Jinzhou Medical University, Jinzhou, China
| | - Qiong Wang
- Department of Ultrasound, the Fourth Medical Center of PLA General Hospital, Beijing, China
| | - Huixia Zhao
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Wenhua Xiao
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Liang Peng
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Jingwen Yang
- Department of Oncology, Senior Department of Oncology, the Fifth Medical Center of PLA General Hospital, Beijing, China.
| | - Xiaoling Yu
- Department of Interventional Ultrasound, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, 100071, China.
| |
Collapse
|
6
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
7
|
Wang Y, Gao L, Cao Y, Yan D, Lukman R, Zhang J, Li Q, Liu J, Du F, Zhang L. Research progress on the synthesis, performance regulation, and applications of Prussian blue nanozymes. Int J Biol Macromol 2025; 295:139535. [PMID: 39761892 DOI: 10.1016/j.ijbiomac.2025.139535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/14/2025]
Abstract
Nanocatalytic medicine offers a novel solution to address the issues of low efficacy, potential side effects, and the development of drug resistance associated with traditional therapies. Therefore, developing highly efficient and durable nanozymes is of great significance for treating diseases related to oxidative stress. In recent years, prussian blue nanoparticles (PBNPs) have been demonstrated to possess multiple enzyme-like catalytic activities and are thus referred to as prussian blue nanozymes (PBNZs). Their excellent biocompatibility and biodegradability make PBNZs promising candidates as biomedical materials. Due to their remarkable catalytic activities, PBNZs have shown great potential in various biomedical applications, such as heavy metal detoxification, antioxidative damage, and anticancer therapies. This paper systematically summarizes the Synthetic strategies of PBNZs, analyzes the regulatory factors of their catalytic performance, and highlights the corresponding modulation methods. Furthermore, the biomedical applications of PBNZs are also reviewed. This study aims to provide researchers with insights and inspirations for the design and preparation of high-performance PBNZs.
Collapse
Affiliation(s)
- Yiyang Wang
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Lei Gao
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Yue Cao
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Duanfeng Yan
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Rilwanu Lukman
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Jingxi Zhang
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Quan Li
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Jiaying Liu
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China
| | - Fengyi Du
- Jiangsu Key Laboratory of Laboratory Medicine, School of medicine, Jiangsu University, Zhenjiang 212013, PR China; Department of Ultrasound, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, PR China
| | - Li Zhang
- Department of Critical Care Medicine Unit, Shanghai Baoshan District Wusong Central Hospital (Zhongshan Hospital Wusong Branch, Fudan University), Shanghai 201900, PR China.
| |
Collapse
|
8
|
Qu J, Cai Y, Li F, Li X, Liu R. Potential therapeutic strategies for colitis and colon cancer: bidirectional targeting STING pathway. EBioMedicine 2025; 111:105491. [PMID: 39644772 PMCID: PMC11665664 DOI: 10.1016/j.ebiom.2024.105491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024] Open
Abstract
The cyclic-GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) pathway has emerged as a promising therapeutic target for colitis and colon cancers. Notably, inhibiting STING may mitigate the progression of colitis, whereas activating STING can enhance anti-tumor immune responses against colon cancer. This duality suggests that the development of STING agonists and inhibitors possesses significant clinical translational potential. In this review, we provide a comprehensive overview of STING inhibitors/agonists that have been systematically evaluated in the contexts of colitis and colon cancer and their specific molecular mechanisms. Other well-characterized STING inhibitors/agonists may also hold considerable promise for the treatment of these conditions, although efficacy validation remain necessary. Additionally, we delve into the latest advances concerning STING oligomerization, degradation and phase separation-dependent self-regulation, proposing potential druggable targets that could inspire the development of novel STING agonists or inhibitors. In Summary, targeting STING appears to be a promising strategy for the treatment of colitis and colon cancer.
Collapse
Affiliation(s)
- Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Yajie Cai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Fanghong Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China.
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 11 Bei San Huan Dong Lu, Beijing, 100029, China.
| |
Collapse
|
9
|
Liu C, Tang L, Yang W, Gu Y, Xu W, Liang Z, Jiang J. cGAS/STING pathway and gastrointestinal cancer: Mechanisms and diagnostic and therapeutic targets (Review). Oncol Rep 2025; 53:15. [PMID: 39611480 PMCID: PMC11632663 DOI: 10.3892/or.2024.8848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
The health of individuals is seriously threatened by intestinal cancer, which includes pancreatic, colorectal, esophageal, gastric and gallbladder cancer. Most gastrointestinal cancers do not have typical and specific early symptoms, and lack specific and effective diagnostic markers and treatment methods. It is critical to understand the etiology of gastrointestinal cancer and develop more efficient methods of diagnosis and treatment. The cyclic GMP‑AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway serves a crucial role in the occurrence, progression and treatment of gastrointestinal cancer. The present review focuses on the latest progress regarding the role and mechanism of the cGAS/STING pathway in gastrointestinal cancer, and discusses treatment approaches and related applications based on the cGAS/STING signaling pathway. In order to improve the knowledge of the connection between the cGAS/STING pathway and gastrointestinal cancer, aid the diagnosis and treatment of gastrointestinal cancer, and lessen the burden on patients and society, the present review also discusses future research directions and existing challenges regarding cGAS/STING in the study of gastrointestinal cancer.
Collapse
Affiliation(s)
- Chang Liu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Li Tang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenhui Yang
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yuning Gu
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wenrong Xu
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhaofeng Liang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, Jiangsu 215600, P.R. China
| |
Collapse
|
10
|
Li X, Liu Y, Ke J, Wang Z, Han M, Wang N, Miao Q, Shao B, Zhou D, Yan F, Ji B. Enhancing Radiofrequency Ablation for Hepatocellular Carcinoma: Nano-Epidrug Effects on Immune Modulation and Antigenicity Restoration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2414365. [PMID: 39548919 DOI: 10.1002/adma.202414365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/25/2024] [Indexed: 11/18/2024]
Abstract
Radiofrequency ablation (RFA), a critical therapy for hepatocellular carcinoma (HCC), carries a significant risk of recurrence and metastasis, particularly owing to mechanisms involving immune evasion and antigen downregulation via epigenetic modifications. This study introduces a "nano-epidrug" named MFMP. MFMP, which is composed of hollow mesoporous manganese dioxide (MnO2) nanoparticles, FIDAS-5 as an MAT2A inhibitor, macrophage membrane, and anti-PD-L1 (aPD-L1), targets HCC cells. By selectively binding to these cells, MFMP initially reverses immune suppression via PD-L1 inhibition. After endocytosis, MFMP disassembles in the tumor microenvironment, releasing FIDAS-5 and Mn2+. FIDAS-5 prevents cGAS methylation, whereas Mn2+ aids STING pathway restoration. In addition, FIDAS-5 reduces m6A RNA modification, suppressing EGFR expression. These changes enhance HCC antigenicity to promote cytotoxic T cell recognition and cytotoxic killing. Furthermore, MFMP mediates immunogenic cell death in HCC by synergizing with RFA through cGAS DNA demethylation, EGFR mRNA demethylation, and TBK1 protein phosphorylation, thereby inhibiting recurrence and metastasis and enhancing immune memory. Thus, MFMP is a potential adjunctive therapy requiring clinical validation.
Collapse
Affiliation(s)
- Xiaocheng Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Jianji Ke
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Zhihua Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Mingda Han
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Ning Wang
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Qiannan Miao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Bingru Shao
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Dan Zhou
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| | - Fei Yan
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun, 130012, China
| | - Bai Ji
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, 130021, China
| |
Collapse
|
11
|
Zhang WX, Li WY, Shu Y, Wang JH. Manganese-enriched prussian blue nanohybrids with smaller electrode potential and lower charge transfer resistance to enhance combination therapy. Colloids Surf B Biointerfaces 2024; 241:114045. [PMID: 38897024 DOI: 10.1016/j.colsurfb.2024.114045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/04/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Prussian blue (PB) is authenticated in clinical treatment, while it generally exhibits unfavorable chemodynamic therapy (CDT) performance. Herein, we developed manganese-doped prussian blue (PBM) nanoparticles to significantly enhance both CDT and photothermal therapy (PTT) effect. The lower redox potential of Mn3+/2+ (0.088 V) in PBM against that of Fe2+/3+ (0.192 V) in PB leads to favorable electron transfer of PBM with respect to PB. Besides, PBM has a lower charge-transfer resistance (Rct) of 2.98 Ω than 4.83 Ω of PB. Once PBM entering the tumor microenvironment (TME), Mn3+ may be readily reduced by glutathione (GSH) and therein to enhance intracellular oxidative stress. Meanwhile, the superoxide dismutase (SOD)-like activity of PBM facilitates the conversion of endogenous superoxide (O2•-) into H2O2. Mn2+ subsequently catalyzes H2O2 to generate toxic hydroxyl radicals (•OH). Notably, the PBM plus laser irradiation can effectively trigger a robust immunogenic cell death (ICD) due to the combination therapy of CDT and PTT. Additionally, the mice treated by PBM followed by laser irradiation efficiently avoided splenomegaly and lung metastasis, along with significant up-regulation of the Stimulator of Interferon Genes (STING) expression. Overall, PBM significantly inhibits tumor growth and metastasis, making it a promising multifunctional nanoplatform for cancer treatment.
Collapse
Affiliation(s)
- Wen-Xin Zhang
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Wang-Yang Li
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China
| | - Yang Shu
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
| | - Jian-Hua Wang
- Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang 110819, China.
| |
Collapse
|
12
|
Ma K, Chen Z, Liang K, Pei Y, Pei Z. A near-infrared light-driven Janus nanomotor for deep tumor penetration and enhanced tumor immunotherapy. Chem Commun (Camb) 2024; 60:9550-9553. [PMID: 39150078 DOI: 10.1039/d4cc03445g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
A near-infrared light-driven Janus nanomotor is constructed by collagenase-coated gold nanorods and chitosan-functionalized mesoporous organosilica nanoparticles with Mn2+ as the bridging ion. The nanomotors with excellent motility and collagenase activity can potently penetrate into tumors to sufficiently activate innate immune responses, significantly enhancing anti-tumor immune efficacy in vivo.
Collapse
Affiliation(s)
- Ke Ma
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, P. R. China.
| | - Zelong Chen
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, P. R. China.
| | - Kai Liang
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, P. R. China.
| | - Yuxin Pei
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, P. R. China.
| | - Zhichao Pei
- Shaanxi Key Laboratory of Natural Products & Chemical Biology, College of Chemistry & Pharmacy, Northwest A&F University, Yangling 712100, P. R. China.
| |
Collapse
|
13
|
Fang K, Zhang H, Kong Q, Ma Y, Xiong T, Qin T, Li S, Zhu X. Recent Progress in Photothermal, Photodynamic and Sonodynamic Cancer Therapy: Through the cGAS-STING Pathway to Efficacy-Enhancing Strategies. Molecules 2024; 29:3704. [PMID: 39125107 PMCID: PMC11314065 DOI: 10.3390/molecules29153704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Photothermal, photodynamic and sonodynamic cancer therapies offer opportunities for precise tumor ablation and reduce side effects. The cyclic guanylate adenylate synthase-stimulator of interferon genes (cGAS-STING) pathway has been considered a potential target to stimulate the immune system in patients and achieve a sustained immune response. Combining photothermal, photodynamic and sonodynamic therapies with cGAS-STING agonists represents a newly developed cancer treatment demonstrating noticeable innovation in its impact on the immune system. Recent reviews have concentrated on diverse materials and their function in cancer therapy. In this review, we focus on the molecular mechanism of photothermal, photodynamic and sonodynamic cancer therapies and the connected role of cGAS-STING agonists in treating cancer.
Collapse
Affiliation(s)
- Kelan Fang
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Huiling Zhang
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- Department of Medicine and Pharmacy, Shizhen College of Guizhou University of Traditional Chinese Medicine, Guiyang 550000, China
| | - Qinghong Kong
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Yunli Ma
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
| | - Tianchan Xiong
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Tengyao Qin
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Sanhua Li
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
| | - Xinting Zhu
- Guizhou Provincial College-Based Key Lab for Tumor Prevention and Treatment with Distinctive Medicines, Zunyi Medical University, Zunyi 563000, China
- College of Basic Medicine, Zunyi Medical University, Zunyi 563000, China
| |
Collapse
|
14
|
Liu L, Zhao X. Preparation of environmentally responsive PDA&DOX@LAC live drug carrier for synergistic tumor therapy. Sci Rep 2024; 14:15927. [PMID: 38987493 PMCID: PMC11236969 DOI: 10.1038/s41598-024-66966-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/05/2024] [Indexed: 07/12/2024] Open
Abstract
The development of intelligent, environmentally responsive and biocompatible photothermal system holds significant importance for the photothermal combined therapy of tumors. In this study, inspired by Lactobacillus (LAC), we prepared a biomimetic nanoplatform PDA&DOX@LAC for tumor photothermal-chemotherapy by integrating the chemotherapeutic drug doxorubicin (DOX) with dopamine through oxidative polymerization to form polydopamine (PDA) on the surface of LAC. The PDA&DOX@LAC nanoplatform not only achieves precise and controlled release of DOX based on the slightly acidic microenvironment of tumor tissues, but also exhibits enzyme-like properties to alleviate tumor hypoxia. Under near-infrared light irradiation, it effectively induces photothermal ablation of tumor cells, enhances cellular uptake of DOX with increasing temperature, and thus efficiently inhibits tumor cell growth. Moreover, it is further confirmed in vivo experiments that photothermal therapy combined with PDA&DOX@LAC induces tumor cells apoptosis, releases tumor-associated antigens, which is engulfed by dendritic cells to activate cytotoxic T lymphocytes, thereby effectively suppressing tumor growth and prolonging the survival period of 4T1 tumor-bearing mice. Therefore, the PDA&DOX@LAC nanoplatform holds immense potential in precise tumor targeting as well as photothermal combined therapy and provides valuable insights and theoretical foundations for the development of novel tumor treatment strategies based on endogenous substances within the body.
Collapse
Affiliation(s)
- Lu Liu
- The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, No. 62, Huaihai Road (S.), Huai'an, 223002, People's Republic of China
| | - Xuefen Zhao
- Northern Jiangsu People's Hospital, Yangzhou, 225001, People's Republic of China.
| |
Collapse
|
15
|
Yang H, Yang S, Guo Q, Sheng J, Mao Z. ATP-Responsive Manganese-Based Bacterial Materials Synergistically Activate the cGAS-STING Pathway for Tumor Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310189. [PMID: 38414097 DOI: 10.1002/adma.202310189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/10/2024] [Indexed: 02/29/2024]
Abstract
Stimulating the cyclic guanosine monophophate(GMP)-adenosine monophosphate (AMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway is a crucial strategy by which bacteria activate the tumor immune system. However, the limited stimulation capability poses significant challenges in advancing bacterial immunotherapy. Here, an adenosine 5'-triphosphate (ATP)-responsive manganese (Mn)-based bacterial material (E. coli@PDMC-PEG (polyethylene glycol)) is engineered successfully, which exhibits an exceptional ability to synergistically activate the cGAS-STING pathway. In the tumor microenvironment, which is characterized by elevated ATP levels, this biohybrid material degrades, resulting in the release of divalent manganese ions (Mn2+) and subsequent bacteria exposure. This combination synergistically activates the cGAS-STING pathway, as Mn2+ enhances the sensitivity of cGAS to the extracellular DNA (eDNA) secreted by the bacteria. The results of the in vivo experiments demonstrate that the biohybrid materials E. coli@PDMC-PEG and VNP20009@PDMC-PEG effectively inhibit the growth of subcutaneous melanoma in mice and in situ liver cancer in rabbits. Valuable insights for the development of bacteria-based tumor immunotherapy are provided here.
Collapse
Affiliation(s)
- Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310003, China
| | - Sisi Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Quanshi Guo
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310003, China
| | - Jifang Sheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Department of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhengwei Mao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310003, China
| |
Collapse
|
16
|
Cao Y, Li Y, Ren C, Yang C, Hao R, Mu T. Manganese-based nanomaterials promote synergistic photo-immunotherapy: green synthesis, underlying mechanisms, and multiple applications. J Mater Chem B 2024; 12:4097-4117. [PMID: 38587869 DOI: 10.1039/d3tb02844e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Single phototherapy and immunotherapy have individually made great achievements in tumor treatment. However, monotherapy has difficulty in balancing accuracy and efficiency. Combining phototherapy with immunotherapy can realize the growth inhibition of distal metastatic tumors and enable the remote monitoring of tumor treatment. The development of nanomaterials with photo-responsiveness and anti-tumor immunity activation ability is crucial for achieving photo-immunotherapy. As immune adjuvants, photosensitizers and photothermal agents, manganese-based nanoparticles (Mn-based NPs) have become a research hotspot owing to their multiple ways of anti-tumor immunity regulation, photothermal conversion and multimodal imaging. However, systematic studies on the synergistic photo-immunotherapy applications of Mn-based NPs are still limited; especially, the green synthesis and mechanism of Mn-based NPs applied in immunotherapy are rarely comprehensively discussed. In this review, the synthesis strategies and function of Mn-based NPs in immunotherapy are first introduced. Next, the different mechanisms and leading applications of Mn-based NPs in immunotherapy are reviewed. In addition, the advantages of Mn-based NPs in synergistic photo-immunotherapy are highlighted. Finally, the challenges and research focus of Mn-based NPs in combination therapy are discussed, which might provide guidance for future personalized cancer therapy.
Collapse
Affiliation(s)
- Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Yilin Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Caixia Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Chengkai Yang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P. R. China
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, P. R. China.
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Tiancheng Mu
- Department of Chemistry, Renmin University of China, Beijing 100872, P. R. China.
| |
Collapse
|
17
|
Cui M, Tang D, Wang B, Zhang H, Liang G, Xiao H. Bioorthogonal Guided Activation of cGAS-STING by AIE Photosensitizer Nanoparticles for Targeted Tumor Therapy and Imaging. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305668. [PMID: 37668998 DOI: 10.1002/adma.202305668] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/30/2023] [Indexed: 09/06/2023]
Abstract
Photodynamic therapy (PDT) and photothermal therapy (PTT) leverage reactive oxygen species (ROS) and control local hyperthermia by photosensitizer to perturb intracellular redox equilibrium, inducing DNA damage in both mitochondria and nucleus, activating the cGAS-STING pathway, ultimately eliciting antitumor immune responses. However, current photosensitizers are encumbered by limitations such as suboptimal tumor targeting, aggregation-caused quenching (ACQ), and restricted excitation and emission wavelengths. Here, this work designs novel nanoparticles based on aggregation-induced emission (AIE) photosensitizer (BODTPE) for targeted tumor therapy and near-infrared II fluorescence imaging (NIR-II FLI) with enhanced PDT/PTT effects. BODTPE is employed as a monomer, dibenzocyclooctyne (DBCO)-PEG2k -amine serving as an end-capping polymer, to synthesize a BODTPE-containing polymer (DBD). Further, through self-assembly, DBD and mPEG-DSPE2k combined to form nanoparticles (NP-DBD). Notably, the DBCO on the surface of NP-DBD can react with azide groups on cancer cells pretreated with Ac4 ManNAz through a copper-free click reaction. This innovative formulation led to targeted accumulation of NP-DBD within tumor sites, a phenomenon convincingly demonstrated in murine tumor models subjected to N-azidoacetylmannosamine-tetraacylated (Ac4 ManNAz) pretreatment. Significantly, NP-DBD exhibits a multifaceted effect encompassing PDT/PTT/NIR-II FLI upon 808 nm laser irradiation, thereby better activating the cGAS-STING pathway, culminating in a compelling tumor inhibition effect augmented by robust immune modulation.
Collapse
Affiliation(s)
- Minhui Cui
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Dongsheng Tang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Bin Wang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hanchen Zhang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Ganghao Liang
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, Laboratory of Polymer Physics and Chemistry, Institute of Chemistry Chinese Academy of Sciences, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
18
|
Hu R, Chen X, Li Z, Zhao G, Ding L, Chen L, Dai C, Chen Y, Zhang B. Liquid Nanoparticles for Nanocatalytic Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2306469. [PMID: 37669827 DOI: 10.1002/adma.202306469] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/31/2023] [Indexed: 09/07/2023]
Abstract
Nanotechnology is revolutionizing cancer therapy, and catalyzes the emerging of ion-involved cancer-therapeutic modality, which unfortunately suffers from undesirable nanocarriers for efficient intracellular ion delivery. To radically extricate from this critical issue, the glutathione (GSH)-responsive organosilica network is employed to lock the liquid drops at the nanoscale via a general bottom-up strategy to achieve the systemic delivery of "ion drugs". In this work, a sulfate radical generation donor (Na2 S2 O8 ), as a paradigm "ion drug", is entrapped into this liquid nanoparticle for efficiently delivering to the tumor region. After further surface engineering with pH-responsive tannic acid-Fe2+ framework, these liquid nanoparticles achieve tumor-microenvironmental pH/GSH-dual responsive ion release (Fe2+ /Na+ /S2 O8 2- ) after reaching the tumor sites, where the Fe2+ further triggers S2 O8 2- to generate toxic •SO4 - and •OH, effectively executing cancer cell ferroptosis (Fe2+ , reactive oxygen species-ROS) and pyroptosis (Na+ , ROS). Such a tumor-responsive/specific liquid nanoplatform is highly instructive for further ion-mediated nanomedicine and disease treatment.
Collapse
Affiliation(s)
- Ruizhi Hu
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P. R. China
| | - Xiaoying Chen
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P. R. China
| | - Zhifang Li
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Guojun Zhao
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, P. R. China
| | - Li Ding
- Education Institute, Tongji University School of Medicine, Tongji University Cancer Center, Shanghai Engineering Research Center of Ultrasound Diagnosis and Treatment, National Clinical Research Center of Interventional Medicine, Shanghai, 200072, P. R. China
| | - Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Chen Dai
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute of Shanghai University, Wenzhou, Zhejiang, 325088, P. R. China
| | - Bo Zhang
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, P. R. China
| |
Collapse
|
19
|
Yang J, Guo W, Huang R, Bian J, Zhang S, Wei T, He C, Hu Z, Li J, Zhou C, Lu M. Self-assembled albumin nanoparticles induce pyroptosis for photodynamic/photothermal/immuno synergistic therapies in triple-negative breast cancer. Front Immunol 2023; 14:1173487. [PMID: 37342347 PMCID: PMC10279487 DOI: 10.3389/fimmu.2023.1173487] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/12/2023] [Indexed: 06/22/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a high degree of malignancy, early metastasis, limited treatment, and poor prognosis. Immunotherapy, as a new and most promising treatment for cancer, has limited efficacy in TNBC because of the immunosuppressive tumor microenvironment (TME). Inducing pyroptosis and activating the cyclic guanosine monophosphate-adenosine monophosphate synthase/interferon gene stimulator (cGAS/STING) signaling pathway to upregulate innate immunity have become an emerging strategy for enhancing tumor immunotherapy. In this study, albumin nanospheres were constructed with photosensitizer-IR780 encapsulated in the core and cGAS-STING agonists/H2S producer-ZnS loaded on the shell (named IR780-ZnS@HSA). In vitro, IR780-ZnS@HSA produced photothermal therapy (PTT) and photodynamic therapy (PDT) effects. In addition, it stimulated immunogenic cell death (ICD) and activated pyroptosis in tumor cells via the caspase-3-GSDME signaling pathway. IR780-ZnS@HSA also activated the cGAS-STING signaling pathway. The two pathways synergistically boost immune response. In vivo, IR780-ZnS@HSA + laser significantly inhibited tumor growth in 4T1 tumor-bearing mice and triggered an immune response, improving the efficacy of the anti-APD-L1 antibody (aPD-L1). In conclusion, IR780-ZnS@HSA, as a novel inducer of pyroptosis, can significantly inhibit tumor growth and improve the efficacy of aPD-L1.
Collapse
Affiliation(s)
- Jianquan Yang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Wen Guo
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Rong Huang
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jiaojiao Bian
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Siqi Zhang
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ting Wei
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Chuanshi He
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Ziyue Hu
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Juan Li
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Chunyang Zhou
- Institute of Materia Medica, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Man Lu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Department of Ultrasound Medical Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
20
|
Li Y, Li X, Yi J, Cao Y, Qin Z, Zhong Z, Yang W. Nanoparticle-Mediated STING Activation for Cancer Immunotherapy. Adv Healthc Mater 2023:e2300260. [PMID: 36905358 DOI: 10.1002/adhm.202300260] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/02/2023] [Indexed: 03/12/2023]
Abstract
As the first line of host defense against pathogenic infections, innate immunity plays a key role in antitumor immunotherapy. The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) (cGAS-STING) pathway has attracted much attention because of the secretion of various proinflammatory cytokines and chemokines. Many STING agonists have been identified and applied into preclinical or clinical trials for cancer immunotherapy. However, the fast excretion, low bioavailability, nonspecificity, and adverse effects of the small molecule STING agonists limit their therapeutic efficacy and in vivo application. Nanodelivery systems with appropriate size, charge, and surface modification are capable of addressing these dilemmas. In this review, the mechanism of the cGAS-STING pathway is discussed and the STING agonists, focusing on nanoparticle-mediated STING therapy and combined therapy for cancers, are summarized. Finally, the future direction and challenges of nano-STING therapy are expounded, emphasizing the pivotal scientific problems and technical bottlenecks and hoping to provide general guidance for its clinical application.
Collapse
Affiliation(s)
- Yongjuan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Xinyan Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Jinmeng Yi
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yongjian Cao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450001, China.,The Center of Infection and Immunity, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, 215123, China
| | - Weijing Yang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou, Henan, 450001, China
| |
Collapse
|
21
|
Ponciri Fructus Immatarus Sensitizes the Apoptotic Effect of Hyperthermia Treatment in AGS Gastric Cancer Cells through ROS-Dependent HSP Suppression. Biomedicines 2023; 11:biomedicines11020405. [PMID: 36830941 PMCID: PMC9953356 DOI: 10.3390/biomedicines11020405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Gastric cancer has been associated with a high incidence and mortality, accompanied by a poor prognosis. Given the limited therapeutic options to treat gastric cancer, alternative treatments need to be urgently developed. Hyperthermia therapy is a potentially effective and safe treatment option for cancer; however, certain limitations need to be addressed. We applied 43 °C hyperthermia to AGS gastric cancer cells combined with Ponciri Fructus Immaturus (PF) to establish their synergistic effects. Co-treatment with PF and hyperthermia synergistically suppressed AGS cell proliferation by inducing extrinsic and intrinsic apoptotic pathways. Additionally, PF and hyperthermia suppressed factors related to metastasis. Cell cycle arrest was determined by flow cytometry, revealing that co-treatment induced arrest at the G2/M phase. As reactive oxygen species (ROS) are critical in hyperthermia therapy, we next examined changes in ROS generation. Co-treatment with PF and hyperthermia increased ROS levels, and apoptotic induction mediated by this combination was partially dependent on ROS generation. Furthermore, heat shock factor 1 and heat shock proteins (HSPs) were notably suppressed following co-treatment with PF and hyperthermia. The HSP-regulating effect was also dependent on ROS generation. Overall, these findings suggest that co-treatment with PF and hyperthermia could afford a promising anticancer therapy for gastric cancer.
Collapse
|